Download vaccines

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Duffy antigen system wikipedia , lookup

Antibody wikipedia , lookup

Vaccine wikipedia , lookup

ELISA wikipedia , lookup

Immune system wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Molecular mimicry wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Adaptive immune system wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Innate immune system wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Vaccination wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Immunocontraception wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Immunomics wikipedia , lookup

DNA vaccination wikipedia , lookup

Transcript
Vaccinces 2013, 1, 415-443; doi:10.3390/vaccines1040415
OPEN ACCESS
vaccines
ISSN 2076-393X
www.mdpi.com/journal/vaccines/
Article
DNA-Encoded Flagellin Activates Toll-Like Receptor 5 (TLR5),
Nod-like Receptor Family CARD Domain-Containing Protein 4
(NRLC4), and Acts as an Epidermal, Systemic, and
Mucosal-Adjuvant
Sanna Nyström 1, Andreas Bråve 2, Tina Falkeborn 3, Claudia Devito 3, Björn Rissiek 1,4,
Daniel X. Johansson 5, Ulf Schröder 6, Satoshi Uematsu 7, Shizuo Akira 8, Jorma Hinkula 3
and Steven E. Applequist 1,*
1
2
3
4
5
6
7
8
Center for Infectious Medicine, F59, Department of Medicine, Karolinska Institutet,
Karolinska University Hospital Huddinge, Stockholm 141 86, Sweden
Department of Preparedness, Swedish Institute for Infectious Disease Control, Stockholm 171 82, Sweden
Division of Molecular Virology, Department of Clinical and Experimental Medicine,
Linköping University, Linköping 581 85, Sweden
Institute of Immunology, University Medical Center Hamburg-Eppendorf,
Hamburg D-20246, Germany
Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Nobels väg 16,
Stockholm 171 77, Sweden
Latour AB, Stockholm 171 21, Sweden
Division of Innate immune regulation, International Research and Development Center for Mucosal
Vaccine, Institute for Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
Department of Host of Defense, Research Institutet for Microbial Diseases, Osaka University,
Osaka 565-0871, Japan
* Author to whom correspondence should be addressed; E-Mail: [email protected];
Tel.: +46-8-5858-9688; Fax: +46-8-746-7637.
Received: 18 July 2013; in revised form: 27 August 2013 / Accepted: 30 August 2013 /
Published: 25 September 2013
Abstract: Eliciting effective immune responses using non-living/replicating DNA vaccines is
a significant challenge. We have previously shown that ballistic dermal plasmid DNA-encoded
flagellin (FliC) promotes humoral as well as cellular immunity to co-delivered antigens.
Here, we observe that a plasmid encoding secreted FliC (pFliC(-gly)) produces flagellin capable
of activating two innate immune receptors known to detect flagellin; Toll-like Receptor 5
(TLR5) and Nod-like Receptor family CARD domain-containing protein 4 (NRLC4). To
Vaccince 2013, 1
416
test the ability of pFliC(-gly) to act as an adjuvant we immunized mice with plasmid
encoding secreted FliC (pFliC(-gly)) and plasmid encoding a model antigen (ovalbumin)
by three different immunization routes representative of dermal, systemic, and mucosal
tissues. By all three routes we observed increases in antigen-specific antibodies in serum as
well as MHC Class I-dependent cellular immune responses when pFliC(-gly) adjuvant was
added. Additionally, we were able to induce mucosal antibody responses and Class II-dependent
cellular immune responses after mucosal vaccination with pFliC(-gly). Humoral immune
responses elicited by heterologus prime-boost immunization with a plasmid encoding HIV-1
from gp160 followed by protein boosting could be enhanced by use of pFliC(-gly). We also
observed enhancement of cross-clade reactive IgA as well as a broadening of B cell epitope
reactivity. These observations indicate that plasmid-encoded secreted flagellin can activate
multiple innate immune responses and function as an adjuvant to non-living/replicating
DNA immunizations. Moreover, the capacity to elicit mucosal immune responses, in
addition to dermal and systemic properties, demonstrates the potential of flagellin to be used
with vaccines designed to be delivered by various routes.
Keywords: adaptive immunity; DNA adjuvant; flagellin; NLRC4; TLR5
Abbreviations: TLR, Toll-like Receptor; NLRC4, Nod-like Receptor family CARD domaincontaining protein 4; Naip5, Neuronal apoptosis inhibitory protein 5; HIV-1, Human
Immunodeficiency Virus-1.
1. Introduction
DNA-vaccines are promising tools with great potential for combating infectious disease. Non-living/
replicating DNA vaccines have several advantages over living viral delivery vectors, such as lower
production costs, increased stability, a higher overall safety profile, and recent evidence indicates that
they can provide humans with protective immunity to viral infection [1]. However, living viral vectors
used in DNA vaccine settings (such as Adenovirus) can still elicit stronger immune responses in
humans than naked DNA. Yet in the case of adenovirus, evidence suggests that they may not promote the
desired immune responses to the recombinant antigen. As results from clinical trials show, the use of a
viral vector can, possibly as a consequence of the anti-vector immunity, potentially even enhance the
risk of infection with certain pathogens [2]. These observations emphasize the critical need to continue
research on methods for adjuvanting minimal, non-living/replicating DNA vaccines.
There are many approaches to improving the efficacy of plasmid DNA vaccines such as
choice of delivery method, modifications of antigen location/stability/presentation, and the use of
immunopotentiators [3]. Here, we investigate a formulation-compatible immunopotentiating adjuvant,
with the potential to activate innate and adaptive immune responses through Toll-like Receptor 5 (TLR5)
and/or possibly Nod-like Receptor (NLR) family members NLRC4 and Naip5 [4]. This approach employs
plasmid DNA encoding a secreted form of flagellin (FliC) from Salmonella typhimurium as an
adjuvant in DNA vaccinations. This adjuvant allows mammalian cells to create an environment of
Vaccince 2013, 1
417
sterile-inflammation, thus mimicking natural infection in a safe manner and promoting adaptive
immune responses to co-delivered DNA-encoded antigens [5]. This approach is unique in that it uses a
plasmid-encoded agonist of innate immune receptors to activate a large variety of molecules capable of
promoting adaptive immunity, unlike many other approaches which use single cytokines or chemokines [3].
A major benefit of this system is that it works without physically linking the antigen to flagellin. This
ensures that the antigen is properly folded and processed and constitutes a major practical advantage as the
system is flexible and can be applied with ease to various antigens without the need for time-consuming
development of fusion-constructs. Recombinant flagellin produced in bacteria is currently being used
by many as an experimental adjuvant to promote humoral and cellular immunity against microbial
pathogens [6–8]. However, the use of flagellin in protein-form presents formulation and stability issues
with non-living/replicating DNA vaccines such as plasmids.
In previous work, we vaccinated mice epidermally, using a gene-gun, with a transmembrane-anchored
form of flagellin (pFliC-Tm) and secreted ovalbumin (pOVA). We observed significant increases in
antigen-specific serum IgG levels compared to pOVA alone as well as strong antigen-specific CD4+/8+
cellular immune responses [5]. Importantly, we also showed that the pFliC-Tm adjuvant delivered with
a DNA-encoded nucleoprotein gene from Influenza A resulted in a strong antigen-specific CD4+/8+
cellular immune response which correlated with protection from lethal virus infection [5]. This work
demonstrated that pFliC-Tm acts as an adjuvant when delivered dermally however it is not clear
whether this is the optimal route for eliciting the broadest or strongest immune responses. Additionally,
not all DNA vaccination approaches are applied dermally therefore further studies of adjuvant effects
induced by various delivery routes are warranted.
The HIV-1 pandemic has been estimated to have according with WHO/UNAIDS reports been spread
globally and infected individuals exist in all countries in the world. So far, only a few experimental vaccine
studies have shown promising and protective results in clinical trials. Thus there is a continued need to find
more efficient vaccination strategies to provide protective immunity against the infection. Since, the main
route of infection is via sexual transmission and via mucosal transmission such as breast-feeding, a vaccine
that can provide mucosal immunity would be desirable. However, mucosal vaccines against infectious
disease are few, and only polio, influenza, rotavirus, S. typhi, and V. cholerae have commercially
available vaccines [9]. There are numerous adjuvants now found to promote mucosal immune responses,
some of them lipid based, however none of these are in themselves plasmid-DNA based technologies [10].
Here, we studied if a plasmid vector expressing secreted FliC (pFliC(-gly)) activates TLR5- and
NLRC4/Naip5-specific innate immune responses and acts as an adjuvant to plasmid-encoded antigen by
three different routes representative of dermal, systemic, and mucosal locations. Additionally, we
performed intranasal mucosal immunizations using plasmid encoding the clinically relevant HIV-1
antigen gp160 followed by recombinant HIV-1 protein booster. The ability of pFliC adjuvant to enhance
HIV-1 gp160 envelope immune responses at mucosal and systemic compartments was also investigated.
Vaccince 2013, 1
418
2. Experimental
2.1. Cloning of Vaccination Expression Constructs and Molecular Mechanism Constructs
pOVA and pFliC-Tm(-gly) have been described previously [5]. pFliC-Tm(-gly) was subjected to
site-directed mutagenesis to insert two translational stop-codons after AA 459 of FliC(-gly) (AA
numbering is based on GenBank Accession #D13689). Changes of all constructs were confirmed by
DNA sequencing. Variants of FliC(-gly) were created for testing the molecular mechanism of FliC(-gly)
detection in vitro. First, to make a cytoplasmically-expressed FliC(-gly) the secretion leader sequence
of FliC(-gly) was removed from the pFliC(-gly) vector by PCR amplification using the primers
5'-CCAGGTTCCAATCTTATGTATCCATATGATGTTCCAGATTATGCT-3' and 5'-GCAGCCGCG
GATCCCGGGGTACCTATCGCAGTAAAGAGAGGACGTTTTGCGG-3' with the pFliC(-gly) template
encoding a starting methionine, HA-tag, and complete FliC(-gly) open-reading frame (ORF) without
the secretion leader sequence. Full-length products were digested with Hind III/BamH I and ligated into
pcDNA 3.1/Zeo(+) prepared with HindIII/BamHI to make pcFliC(-gly). To remove the COOH-terminal
34 amino-acids of FliC a section of the FliC(-gly) gene encoding AA282 to 461 residing on a BsrG I/Xho I
fragment (5'-ATGTACAAGTTGCAAATGCTGATTTGACAGAGGCTAAAGCCGCATTGACAGCAG
CAGGTGTTACCGGCACAGCATCTGTTGTTAAGATGTCTTATACTGATAATAACGGTAAAACT
ATTGATGGTGGTTTAGCAGTTAAGGTAGGCGATGATTACTATTCTGCAACTCAAAATAAAG
ATGGTTCCATAAGTATTGATACTACGAAATACACTGCAGATGACGGTACATCCAAAACTGC
ACTAAACAAACTGGGTGGCGCCGACGGCAAAACCGAAGTTGTTTCTATTGGTGGTAAAACT
TACGCTGCAAGTAAAGCCGAAGGTCACAACTTTAAAGCACAGCCTGATCTGGCGGAAGCG
GCTGCTACAACCACCGAAAACCCGCTGCAGAAAATTGATGCTGCTTTGGCACAGGTTGACA
CGTTACGTTCTGACCTGGGTGCGGTACAGAACCGTTTCAACTCCGCTATTACCAACCTGGG
CAACACCGTAAACAACCTGAATTCTGCCCGTAGCCGTATCGAAGATTCCGACTACGCGACC
TAGTAGCTCGAGA-3') was synthesized (and used to replace the 3' end of both the pFliC(-gly) and
pcFliC(-gly) constructs after digestion by BsrG I/Xho I to create pFliC(-gly)34 and pcFliC(-gly)34.
pFliC(-gly), pcFliC(-gly), pFliC(-gly)34, or pcFliC(-gly)34 were transiently transfected into
293T cells and 2 days later cell lysates were prepared as described [5], total protein concentration was
determined by BCA assay (Pierce Thermo Scientific, Walthman, MA, USA), normalized, and
subjected to SDS-PAGE (NuPAGE, Invitrogen Life Technologies, Stockholm, Sweden) followed by
western blot analysis (anti-HA tag, Covance Research Products, Brussels, Belgium).
The ORF of FliC(-gly) gene and three variants were excised using MfeI and XhoI and inserted into
the retroviral expression vector pMSCV-IRES-GFP/neo digested with EcoR I and Xho I. Constructs
were transfected into 293T cells and proteins from cell lysates and supernatants were analyzed for the
presence and correct molecular weight of FliC(-gly) and variants by Western blotting as described
above (data not shown).
Plasmid vectors expressing gp160 and p24gag from HIV-1 clade B strain Ba-L have been described
previously [11].
Vaccince 2013, 1
419
2.2. In Vitro Macrophage Stimulations and Retroviral Lethality Screen
All plasmid DNAs were prepared using a Qiagen EndoFree Plasmid Maxi Kit (Qiagen, Hamburg,
Germany). Macrophage stimulations were performed as follows. Alveolar macrophages were
harvested by BAL from C57BL6/N or TLR5−/− mice backcrossed >10 generations onto C57BL6. Cells
were seeded 50,000 cells/well in 96 well plates (Costar) in 50 µL RPMI 1640, 10% heat-inactivated
FCS, 2 mM L-glutamine, 100 U/mL penicillin and 100 µg/mL streptomycin (Pierce Thermo Scientific,
Walthman, MA, USA) and allowed to settle for 1 h. 50 µL of two day culture supernatants from 293T
cells, transiently transfected with 0.8 µg of pFliC(-gly), pFliC(-gly)34, empty vector, recombinant
FliC at 100 ng/mL (a gift from A. Gewirtz, Emory, GA, USA) or ultra-pure LPS at 100 ng/mL
(InVivogen, San Diego, CA, USA), was placed on cells and incubated at 37 °C at 10% CO2 for 4 h.
Cell supernatants were harvested and subjected to standard ELISA to detect secreted mouse TNF
(BioLegend, San Diego, CA, USA).
Retroviral Lethality Screen was performed as follows. pMSCV-IRES-GFP/neo alone or containing
FliC(-gly), cFliC(-gly), FliC(-gly)34, or cFliC(-gly)34 ORF were packaged in Phoenix amphotropic
virus packaging cells. After transfection media was replaced at day one and viral supernatants were
harvested at days two and three. Pooled supernatants were 0.45 µm filtered, concentrated by centrifugation
at 2,300 × g for 18 h at +4 °C, and frozen at −80 °C. 2 × 105 J2-virus immortalized mouse bone-marrow
derived macrophage cells (BcgR) or 293T cells were pre-treated with BX795 (5 µM) for 30 min at 37 °C
to improve transduction efficiency [12] followed by 200 µL virus mixed with polybrene 8 µg/mL (Sigma,
St. Louis, MO, USA) and centrifuged for 45 min at 27 °C. 4 days after triplicate transductions cells
were subjected to analysis for GFP by flow cytometry using a 4-laser LSRII-Fortessa with standard filter
sets (BD Bioscience, Stockholm, Sweden). >40,000 non-debris singlets were analyzed in every
sample. FACS data was analyzed using FlowJo v9.2 (Tree Star, Ashland, OR, USA).
2.3. Mice and Vaccinations
For experiments using OVA antigen female C57BL6/J-crl sub-strain mice (8–12 weeks at priming)
from Charles River Laboratories (Sulzfeld, Germany) were used and housed under standard specific
pathogen-free conditions (Swedish Institute for Infectious Disease Control). All procedures were
reviewed, approved, and performed under both institutional and national guidelines. Plasmid DNA was
prepared using a Qiagen EndoFree Plasmid Maxi Kit (Qiagen, Hamburg, Germany) as described by the
manufacturer without exception. Vaccinations were done in the animal facility at approximately 24 °C
and 60% relative humidity. Mice receiving intramuscular vaccinations were injected with DNA
resuspended in PBS in a total volume of 50 μL in one quadricep. Standard ballistic dermal vaccinations
were performed as described [5]. For intra-nasal vaccinations, plasmid DNA was resuspended in 0.1 M
Tris-HCL, pH 8.0 and placed on ice and mixed with a 1:1 volume of 2% Eurocine cationic N3 lipid
(called N3) to make a final volume of 1% N3 lipid adjuvant (see Table 1 for details). Preparation of N3
was carried out as described [13] and was gently stirred with DNA on ice until homogenous then brought
to room temperature. Mice were briefly anesthetized with Isofluran, placed dorsal side up, and
4 μL/nostril of N3/DNA mixture was applied to each nostril using a standard laboratory pipette. Mice
were gently supported in this position until the mouse revived and attempted to turn over.
Vaccince 2013, 1
420
Table 1. Vaccinated groups for ovalbumin (OVA) experiments.
Group n
1
2
3
4
5
6
7
8
9
10
11
12
13
7
7
8
8
7
8
8
8
8
7
8
8
7
Total
Immunizations
2
2
2
2
2
2
2
2
2
2
2
2
2
a
Route a
ImmunogenpOVA
g.g.
g.g.
g.g.
g.g.
i.m.
i.m.
i.m.
i.m.
i.na.
i.na.
i.na.
i.na.
i.na.
0.5 µg
0.5 µg
0.5 µg
0.5 µg
10 µg
10 µg
10 µg
10 µg
4 µg
4 µg
4 µg
4 µg
4 µg
Adjuvant
pFliC(-gly)
0.1 µg
0.2 µg
0.5 µg
2 µg
5 µg
10 µg
1 µg
2 µg
4 µg
Empty Vector
pcDNA3.1/Zeo(+)
0.5 µg
0.4 µg
0.3 µg
10 µg
8 µg
5 µg
4 µg
4 µg
3 µg
2 µg
-
N3
Lipid
1%
1%
1%
1%
g.g.=gene-gun, i.m.=intra-muscular, i.na.=intra-nasal.
For experiments involving gp160/p24gag, eight to ten-week-old female BALB/c mice were purchased
from Scanbur BK, Sollentuna, Sweden. Six groups (n = 35) were vaccinated with DNA-plasmids
expressing gp160 and p24gag (promoter CMV-IE) as previously described [11] with or without adjuvant
(Table 2). For plasmid-DNA priming, mice were given 10 µg/plasmid dose/mouse as 5 µL/nostril/mouse
with N3 prepared as described above. HIV-1 recombinant protein-boost antigens were gp160 and
p24gag (Protein Sciences Inc., Meriden, CT, USA), containing HIV-1 gp160 LAI and p24gag prepared
with anionic L3B as described [14]. Mice were given 5 µL of vaccine in each nostril, corresponding to
1 µg recombinant protein antigen and 0%, or 2% of L3B adjuvant or 1% N3 with 5 µg pFliC(-gly) (see
Table 2 for details). For delivery of intranasal vaccinations, the mice were treated as above. Groups
studied for longetivity of immune responses were immunized three times at three weeks intervals then
mice were sacrificed 4, 8, 12, 24, and 36 weeks after the last immunization for analysis. Mice studied
for general immune reactivity were sacrificed 4 weeks after the final immunization.
Table 2. Vaccinated groups for gp160/p24gag experiments.
Group
n
Total
Immunizations
1
35
3
2
35
3
3
35
3
4
35
3
Priming
ImmunogenP a
pgp160(5 µg)
pGagp24(5 µg)
pgp160(5 µg)
pGagp24(5 µg)
pgp160(5 µg)
pGagp24(5 µg)
pgp160(5 µg)
pGagp24( 5µg)
Boosting
Adjuvant
EmptyVectorpc
DNA3.1/Zeo(+)
None
-
N3 (1%)
-
pFliC(-gly)
(5 µg)
N3(1%) +
pFliC(gly)
(5 µg)
-
-
ImmunogenB b
Adjuvant
rgp160/rp24gag
(1 µg each)
rgp160/rp24gag
(1 µg each)
rgp160/rp24gag
(1 µg each)
L3B(2%)
rgp160/rp24gag
(1 µg each)
N3(1%) +
pFliC(gly) (5 µg)
None
L3B(2%)
Vaccince 2013, 1
421
Table 2. Cont.
Group
n
Total
Immunizations
5
35
6
Saline
Priming
Boosting
ImmunogenP a
Adjuvant
EmptyVectorpc
DNA3.1/Zeo(+)
3
pgp160(5 µg)
pGagp24(5 µg)
None
5 µg
rgp160/rp24gag
(1 µg each)
35
3
pgp160(5 µg)
pGagp24(5 µg)
N3(1%)
5 µg
rgp160/rp24gag
(1 µg each)
30
3
ImmunogenB b
Adjuvant
N3(1%) +
pcDNA3.1 (5 µg)
N3(1%) +
pcDNA3.1
(5 µg)
Saline
a
Plasmid DNA; b Recombinant protein.
2.4. Antibody, Mucosal Cytokines and T Cell Analysis
Anti-OVA humoral responses were performed as follows. Briefly, serum, fecal pellets (100 mg
feces solublized in 1 mL PBS with protease inhibitors, Complete Mini, Roche, ) and vaginal washings
(50 μL of PBS with protease inhibitors, as above) were subjected to anti-OVA ELISA as described [5].
Assessment of antigen-specific IgA in lungs was done as follows. Isolated lungs were rinsed in cold
PBS then minced in PBS with protease inhibitors. Solids were removed by centrifugation and total IgA in
washings were determined by ELISA, using a primary monoclonal goat-anti mouse IgA (Sigma 098K4823
clone ISO2-1KT, St. Louis, MO, USA) and secondary rabbit anti-goat IgG HRP (Dako, Stockholm,
Sweden). IgA anti-OVA titers were determined by ELISA then normalized for total IgA content.
Individual samples were tested in triplicate at a dilution of 1/10. Amino acids were numbered
according to the Los Alamos Data Base on Retroviruses (peptide source: AIDS Research and
Reference Reagent Program, Division of AIDS, NIAID, NIH: from DAIDS, NIAID and J&J, San Diego,
CA, USA) [15]. For IgG anti-gp160 measurements, individual mouse sera were diluted in ten-fold steps
from a starting dilution of 1:100 in ELISA-buffer (2.5% dry milk and 0.05% Tween-20 (Sigma, St. Louis,
MO, USA) in PBS to end-point. Goat-anti-mouse IgG (H+L)-HRP secondary conjugate (Bio-Rad,
Hercules, CA, USA) was used, diluted 1:3,000 to detect IgG anti-gp160 immune complexes. Anti-gp160
IgA and IgG isotype subclasses were measured using a mouse monoclonal antibody isotyping reagent
(Sigma, St. Louis, MO, USA) according to the manufacturer’s protocol with peroxidase-conjugated
anti-Goat IgG (Sigma, St. Louis, MO, USA), diluted 1:2,000. For developing ELISA reactions,
O-phenylenediaminedihydrochloride (OPD) (Sigma, St. Louis, MO, USA) was used. Based on earlier
studies, an OD of 0.2 was set as the cut-off value for positive samples. Clade A, Uganda 29 (UG29) and C,
Brazil (BR25) envelope antigens were obtained from the AIDS Research and Reference Reagent Program,
Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health
(NIAID, NIH, Bethesda, MD, USA). Mucosal wash IgA analyses were performed as previously
described [16–18]. Briefly, IgA was isolated from secretions collected by nasal washing using
Kaptive/IgA/IgE reagents (Biotech IgG, Copenhagen, Denmark) as recommended by the manufacturer.
IgA quantities were determined using an in-house murine IgA capture ELISA, and commercial murine
IgA (1 mg/mL, Sigma, St. Louis, MO, USA) was used to prepare a standard curve. The purified IgA
and the standard IgA were diluted in ten-fold serial dilutions. From each dilution, 100 µL was added to
each well of a 96-microwell plate pre-coated with rabbit anti-murine IgA (Dakopatts AB, Sollentuna,
Vaccince 2013, 1
422
Sweden). Goat-anti-mouse IgA-HRP secondary conjugate (SouthernBiotech, Birmingham, AL, USA),
diluted by 1:3,000, was used to detect IgA anti-gp160. The total amounts of IgA in nasal samples were
determined by comparing the OD values of the test samples with the IgA standard and final anti-gp160
values were normalized to total IgA values.
T-cell responses to OVA was performed as described [5]. Briefly, spleens were isolated, single-cell
suspensions were Ficoll-purified, washed twice with PBS and used in IFNγ ELISPOT analysis
according to the manufacturer guidelines (Mabtech, Nacka, Sweden). Ag restimulation was performed
using either the H-2Kb binding OVA peptide SIINFEKL (257-264) at 1 μM final concentration or the
I-Ab binding OVA peptide ISQAVHAAHAEINEAGR (323-339) at 1 μM final concentration (GenScript,
Piscataway, NJ, USA).). Cell reactivity was confirmed by incubation with ConA. Spot-forming cells were
quantified after 24 h incubation and counted by AID ELISPOT reader (AutoImmun Diagnostika,
Straßberg, Germany).
T cell immune responses to gp160 were measured using a cell-in-well murine cytokine capture-ELISA
assay as described previously [11]. Briefly, 96-well ELISA plates were coated with capture anti-IFN
(AN18) or anti-IL-5 (TRFK4) according to the manufacturer’s protocol (Mabtech, Nacka, Sweden)
overnight at 4 °C. Following well washing and blocking according to the manufacturer’s protocol 2.5 ×105
ficoll-purified splenocytes from individual mice were added to each well, either with or without
recombinant HIV-1 gp160, p24gag (Protein Sciences Inc., Meriden, CT, USA), control antigen (Sf9
cell lysate), the positive control Concanavalin A (2.5 µg/mL, Sigma, St. Louis, MO, USA) or RPMI
1640-medium alone. Plates were incubated at 37 °C, 5% CO2 for 5–6 days. Cells were then removed,
plates were washed with PBS, and biotinylated detection antibodies were added, washed, followed by
streptavidin-ALP (Mabtech, Nacka, Sweden). The plates were developed with substrate solution
(Mabtech, Nacka, Sweden) for 5–10 min until spots became visible, and the color reaction was stopped
by 2.5 M H2SO4. Plates were then read in an ELISA reader (BioRad, Hercules, CA, USA) at 405 nm.
T-cell proliferation to gp160 was performed as described previously [18] using 1 µg/mL of rgp160
or rGag p24 as specific antigens.
Nasal washings were performed trice with 25 µL PBS/nostril on Isofluran sedated mice kept in
supine position. Collected washings were frozen until analyzed. Nasal washings were tested individually
for the cytokines IL-6, IFN, and IFN according to the manufacturer’s protocols (R&D Systems,
Minneapolis, MN, USA).
2.5. HIV-1 Neutralization Assay
The HIV-1 neutralization assay was performed as described previously [11]. The viral isolates used
for the neutralization were the subtype B laboratory strains IIIB LAI (vaccine homologus) and the
primary subtype B isolate 6,794. Briefly, the sera from mice were pooled group wise and inactivated at
56 °C for 1 h to prevent complement-mediated neutralization. Sera were diluted in RPMI 1640
medium (Invitrogen Life Technologies, Stockholm, Sweden) in 96-well tissue culture plates (Nunc
microwell plates, Nunc, Pierce Thermo Scientific, Walthman, MA, USA). Dilutions were mixed with
virus and incubated at 37 °C for 1 h followed by the addition of 1 × 105 human PBMCs (activated by
phytohemagglutinine and rIL-2; PeproTech, Rocky Hill, NJ, USA) or Jurkat T cells. The cultures were
incubated at 37 °C in 5% CO2 over night, after which the cells were washed twice with RPMI 1640.
Vaccince 2013, 1
423
After 6 days of culture, the presence of HIV-1 p24 antigen in the culture medium was measured by
ELISA [19]. The background in the p24 ELISA was determined for each plate and subtracted from all
wells before the percentage neutralization was determined as [1-(mean p24 OD in the presence of test
serum/mean p24 OD in the absence of test serum)] × 100. Ethical permission for use of huPBMCs was
approved by the ethical committee at Linköping University Hospital.
Statistical analysis was performed using GraphPad Prism 5 (La Jolla, CA, USA). Comparisons
between groups with the HIV-1 antigens were performed by using the non-parametric Mann-Whitney
U test with Bonferroni correction, p < 0.05 was considered significant.
3. Results and Discussion
3.1. Construction of Secreted FliC Adjuvant
A secreted variant of flagellin with reduced glycosylation (called pFliC(-gly)), based on the pFliC-Tm
(-gly) plasmid [5], was constructed by removing the human transmembrane PDGFR domain from the
ORF to eliminate potential immune responses to this region and to prepare a base vector for adjuvant
use. Three additional variants of pFliC(-gly) were also constructed to test the ability of FliC(-gly) to
activate the two known innate immune receptors capable of sensing flagellin TLR5 and NLRC4/Naip5.
These four constructs are depicted in Figure 1a relative to the defined domains of Salmonella
typhimurium FliC. To prepare pFliC(-gly) control variants capable of activating cytoplasmically expressed
NLRC4/Naip5 we recloned the FliC(-gly) insert sans leader sequence (pcFliC(-gly)). We also prepared
additional control versions of pFliC(-gly) and pcFliC(-gly) removing the COOH-34 amino-acids of
FliC(-gly) shown to activate NLRC4/Naip5 [20]. These versions were designated pFliC(-gly)34 and
pcFliC(-gly)34 respectively. All four vectors were capable of expressing proteins of predicted size
with an apparent polypeptide of approximately 52 kDa for pFliC(-gly) and pcFliC(-gly) and
approximately 48 kDa for pFliC(-gly)34 and pcFliC(-gly)34 (Figure 1b). To determine if secreted
FliC(-gly) protein produced from pFliC vectors could activate TLR5 culture supernatants from pFliC(-gly),
pFliC(-gly)34 transfected 293 cells, or recombinant FliC protein were applied to alveolar macrophages
from B6 or TLR5-deficient mice. Plasmid vectors produced full-length or 34 secreted FliC(-gly) able
to activate B6 alveolar macrophages to produce TNF but not macrophages from TLR5-deficient mice
(Figure 1c). To determine if secreted FliC(-gly) has the potential to activate cytoplasmic NLRC4/Naip5
inflammasome responses we performed a retroviral lethality screen using the macrophage cell line BcgR
which undergoes pyroptosis in the presence of the COOH-terminal tail of FliC [21]. This assay detects
the ability of macrophages virally transduced with genes expressing GFP as well as various flagellin
constructs to undergo pyroptotic cell death in response to whole flagellin dependent on the
NLRC4/Naip5 35 amino-acid carboxy-terminal activating domain [20,21]. GFP positive BcgR cells
are taken as evidence of a lack of NLRC4/Naip5 activation while GFP negative cells, relative to GFP
positive identically transduced 293T control cells, are taken as evidence of NLRC4/Naip5 activation.
FliC(-gly), cytoplasmic expressed FliC (cFLiC(-gly)), and a variant of each lacking the final 34 amino-acid
COOH-tail (34) (Figure 1a) were subcloned into the retroviral vector pMSCV-IRES-GFP which are
designed to produce FliC(-gly) and variants as well as GFP upon transduction. Various FliC(-gly)
constructs were Amphotropic packaged, and used to transduce BcgR or 293T cells. Using GFP as a
Vaccince 2013, 1
424
reporter for FliC expression we observed that all versions of FliC(-gly) were expressed at nearly equal
frequency in 293T cells indicating that all vectors were packaged with equal efficiency and could
deliver GFP and FliC genes (Figure 1d). However, when identical vector preparations expressing
FliC(-gly) were trandsuced into BcgR cells we observed GFP expression only with 34 versions
(Figure 1d). These results demonstrate that secreted form of FliC(-gly) we use as an in vivo adjuvant has
the ability to activate NLRC4/Naip5 pyroptotic cell death when expressed in a responsive cell type.
FliC(-gly) produced from pFliC(-gly) stimulated TNF production in a TLR5-dependent manner as
well as inflammatory cell death (pyroptosis) dependent on a defined FliC region known to be a
NLRC4/Naip5 agonist. These results suggest that TLR5 and NLRC4 expressed in vivo could be
important factors in the adjuvant effects of pFliC(-gly) in immunized mice. It is interesting that FliC(-gly)
destined for secretion has the capacity to activate the cytoplasmic flagellin detectors NLRC4/Naip5.
We consider it likely that a portion of secreted FliC(-gly) undergoing translation is retro-translocated
from the endoplasmic reticulum back into the cytoplasm where it may detected by NLRC4/Naip5
leading to the induction of pyroptosis.
3.2. pOVA DNA Vaccinations; Timeline, Routes, and Dose
To compare the effectiveness of secreted flagellin (pFliC(-gly)) as a DNA adjuvant by various routes,
DNA vaccinations were carried out using plasmid pOVA together with empty vector (pcDNA3.1/Zeo(+))
or with vector expressing pFliC(-gly). Empty vector control was used to include possible adjuvant
effects contributed by sensing of B-DNA by innate immune receptors [22,23] but to exclude adjuvant
effects contributed by secreted flagellin (Table 1). Vaccinated mice were primed once, boosted once
and then sampled 9 and 10 days later (Figure 2a). The amount of total DNA given to the mice varied and
was dependent on the limitations of the delivery route (Table 1). Mice were vaccinated by three different
routes representative of dermal (gene-gun, g.g.), systemic (intramuscular, i.m.), and mucosal tissues
(intra-nasal, i.na.). A constant sub-optimal amount of pOVA was used with each route (0.5 μg/g.g.,
10 μg/i.m., 5 μg/i.na.) to allow the study of the adjuvant effects of flagellin.
3.3. Antibody Immune Responses to pOVA DNA Vaccination
To determine if pFliC(-gly) promotes humoral immune response to co-delivered DNA-encoded
antigen (pOVA), we studied antigen-specific antibody responses in dermal, systemic, and mucosal
compartments. When anti-OVA antibody responses were examined in the sera of vaccinated mice we
observed that the pFliC(-gly) adjuvant increased the antigen-specific total IgG antibodies in the sera of
mice vaccinated with pOVA by all three routes (Figure 2b). These responses were dependent on the
dose of pFliC(-gly) used as mice given 0.1 or 0.2 μg of pFliC(-gly) by g.g. or mice given 2 or 5 μg of
pFliC(-gly) by i.m. did not exhibit any increases in anti-OVA antibody responses (data not shown;
Table 1, Groups 2, 3 and 6, 7 respectively). Significant increases were seen when mice were vaccinated
by g.g. or i.na., however, we also observed a reproducible trend of pFliC(-gly) to promote increases in
anti-OVA total IgG when mice were vaccinated i.m. To see if the adjuvant effects of pFliC(-gly)
delivered by various routes affected skewing of anti-OVA IgG isotypes, we analyzed the titers of anti-OVA
IgG1, IgG2b, and IgG2c in the sera of vaccinated mice. We observed increases in all three IgG
isotypes when pFliC(-gly) adjuvant was used, regardless of the route of delivery (Figure 2c–e).
Vaccince 2013, 1
Figure 1. Flagellin (FliC)(-gly) variants for in vivo and in vitro use activate innate immune
responses. (a) Depiction of pFliC(-gly) and variants relative to FliC polypeptide and domains
produced by S. typhimurium. Grey domains D0/1 indicate conserved regions important for
activating innate immune responses. L and HA indicates a leader and HA-epitope tag domains
respectively. Names of the four FliC(-gly) constructs used in this study are indicated to the
right of the drawings; (b) Western blot analysis of cellular lysates from 293T cells transfected
with the indicated constructs. Apparent molecular weights were determined by comparison to
the standard depicted to the left of the blot. Signals were not detected from cells transfected
with empty vector (data not shown); (c) Release of TNF from B6 alveolar macrophages but
not Toll-like Receptor 5 (TLR5) −/− alveolar macrophages after stimulation with FliC(-gly) and
FliC(-gly)34. Supernatant from 293T cells transfected with pFliC(-gly) and pFliC(-gly)34
vectors was incubated with cells for 4 h followed by analysis of secreted TNF by ELISA.
Data are mean ±SEM of triplicate samples representative of two independent experiments; (d)
Activation of pyroptotic cell death by retroviral transduction of BcgR macrophages with
constructs expressing FliC(-gly) but not FliC(-gly)34 as determined by GFP expression. Upper
panels represent representative data from BcgR cells transduced with FliC(-gly), FliC(-gly)34
and controls (as indicated) when comparing GFP and forward-scatter (FS) parameters.
Quantitative data of the percentage of GFP positive BcgR cells from each construct after
transduction. Lower panel represent representative data from 293T cells transduced in identical
fashion. Data are mean ± SEM of GFP positive cells observed during three independent
transduction experiments. * Differences of the response relative to the FliC(-gly) construct
without 34 defined as p ≤ 0.05 calculated using a two-tailed unpaired Student t test.
425
Vaccince 2013, 1
426
Figure 2. Vaccination schedule and serum antibody responses to OVA. (a) Immunization
and sample isolation timeline; (b) Anti-OVA total IgG responses. Anti-OVA IgG1 (c),
IgG2b (d), IgG2c (e) responses. (White bars) g.g. (Dark Grey Bars) i.m. (Grey bars) i.na.
immunized mice. Striped bars indicate the use of pFliC(-gly). Results are representative of
two independent experiments (n = 7–8 mice/group). The concentration of OVA-specific
Abs are expressed as the reciprocal of the last dilution of samples giving an OD equal to, or
higher than, the mean + 3 SDs (the determined cutoff value for the assay) of the values of
serum samples from unimmunized mice. Absorbance values equal to or above the cutoff value
were considered positive. The error bars represent 95% confidence intervals calculated from
the geometric mean titers. * Differences of the response relative to pOVA immunizations
without pFliC(-gly) defined as p ≤ 0.05 were considered significant using a two-tailed
unpaired Student t test.
To assess whether DNA vaccination of mice with pOVA and empty vector or pFliC(-gly) by
various routes could elicit antibody responses in mucosal compartments, we collected extracts from
fecal pellets, vaginal washes, and extracts from lung homogenates. We observed no significant differences
in total amounts of total IgG or IgA immunoglobulins isolated from g.g., i.m. or i.na.-immunized
animals (data not shown). Fecal extract samples were assessed for the relative amount of anti-OVA
total IgG and IgA. We were able to detect significant increases in fecal anti-OVA IgG and IgA in the
Vaccince 2013, 1
427
groups of mice vaccinated intranasally with pOVA together with the highest amounts of pFliC(-gly),
but not when the same plasmids were delivered by g.g. or i.m. (Figure 3a–b). Similarly, only animals
receiving the highest doses of pFliC(-gly) and pOVA intranasally developed measurable anti-OVA
IgA in the vaginal washes (Figure 3c) and lungs (Figure 3d).
We find it interesting that pFliC(-gly) promotes antigen-specific IgG and IgA responses to in mucosal
compartments after mucosal delivery but not when it is delivered systemically or dermally. Despite this
specificity we observed antigen-specific IgG in the sera by all routes. Other studies using purified
flagellin protein and mucosal cell populations however, has revealed that the small intestine lamina
propria contains CD103+ dendritic cells which express TLR5 and respond directly to flagellin to
promote T cell-independent class switching of naive B cells from IgM+IgD+ to IgA [24]. It may be that
a similar phenomenon occurs in vivo when flagellin is present in the compartments of the nasal mucosa
and upper airway. However, it is not known why flagellin acts to promote humoral immune responses
by all routes explored, but does not elicit mucosal antibodies when delivered systemically or dermally.
Differences in the numbers, types, or the immune-skewing potential of flagellin-responsive cells
interacting with flagellin after i.m. or g.g. delivery could be responsible for these effects.
Figure 3. Mucosal antibody responses to OVA. (a) Fecal anti-OVA IgG and (b) IgA
responses; (c) Vaginal anti-OVA IgA responses. (White bars) g.g. (Dark Grey Bars) i.m.
(Grey bars) i.na. immunized mice. Striped bars indicate the use of pFliC(-gly); (d) Lung
anti-OVA IgA responses shown are from mice only vaccinated i.na. and immunizations
given are shown below the axis. Results are representative of two independent experiments
(n = 7–8 mice/group). The concentration of OVA-specific Abs in samples are expressed as
OD equal to, or higher than, the mean OD of the values of samples from unimmunized mice.
The error bars represent SEM calculated from the mean OD. * Differences of the response
relative to pOVA immunizations without pFliC(-gly) defined as p ≤ 0.05 were considered
significant using a two-tailed unpaired Student t test.
Vaccince 2013, 1
428
3.4. Cellular Immune Responses to pOVA DNA Vaccination
MHC class I-dependent responses were analyzed by stimulation of splenocytes from immunized mice
with peptide representing the immunodominant OVA H2-Kb restricted epitope. We observed significant
increases in the numbers of antigen-specific IFNγ-producing cells in mice receiving either g.g. or i.na.
immunization with pOVA and pFliC(-gly) when compared to mice receiving pOVA together with empty
vector (Figure 4a). We also observed a reproducible trend of pFliC(-gly) to promote antigen-specific
increases in IFNγ-producing cells when mice were vaccinated i.m. (Figure 4a). These Class I cellular
responses were dependent on the dose of pFliC(-gly) delivered as mice given 0.1 or 0.2 μg of pFliC(-gly)
by g.g or mice given 2 or 5 μg of pFliC(-gly) i.m. did not exhibit detectable Class I-dependent responses
(data not shown; Table 1, Groups 2, 3 and 6, 7 respectively). When Class II-dependent cellular immune
responses were studied by stimulating splenocytes with the immunodominant I-Ab binding OVA
peptide we observed significant increases in the numbers of antigen-specific IFNγ-producing cells in
mice receiving pOVA intranasally together with the highest amounts of pFliC(-gly), but not with
pOVA and empty vector (Figure 4b). We did not observe any OVA-specific class II-restricted
responses after i.m or g.g. immunization (Figure 4b).
Figure 4. Class I- and Class II-dependent T cell responses to OVA. IFN ELISPOT
analysis of splenic T cell responses to (a) Class-I and (b) Class-II MHC binding OVA
peptides after vaccination. (White bars) g.g. (Dark Grey Bars) i.m. (Grey bars) i.na.
immunized mice. Striped bars indicate the use of pFliC(-gly). Results are representative of
two independent experiments (n = 7–8 mice/group). Data is expressed as the calculated
geometric mean of the Ag-stimulated cells minus unstimulated cells. The error bars represent
SEM calculated from the mean SFC/106 splenocytes. Statistical analyses were conducted using
a two-tailed Student t test. * Differences of the response relative to pOVA immunizations
without pFliC(-gly) defined as p ≤ 0.05 were considered significant using an two-tailed
unpaired student t test.
We observed unique Class-II dependent IFNγ-responses in the spleen after mucosal delivery of
pFliC(-gly) but not when delivered systemically or dermally. This mirrors our observations of mucosal
IgG and IgA with pFliC(-gly) use. Why does pFliC(-gly) promote strong Th1-like CD4+ T cell and
IFNγ-producing CD8+ T cell responses when delivered mucosally but only promotes increases in
IFNγ-producing CD8+ T cells when applied systemically or dermally? It could be that splenic Th1-like
Vaccince 2013, 1
429
CD4+ cells have trafficked to other locations before analysis or are below the level of detection.
Alternatively, there may be a strong ability of FliC to promote CD4+ responses when delivered
mucosally. It has been observed that certain mucosal DC populations expressing TLR5 have a special
ability to promote flagellin-specific CD4+ T cell responses [25]. However, it remains to be seen if these
TLR5-dependent responses can be extended to other antigens encountered in the same environment as
flagellin. Nevertheless, as an adjuvant, we find it interesting that flagellin can promote different immune
responses to the same antigen encountered in different environments. This may have relevance to
immune responses elicited by flagellated pathogens.
Adjuvant effects of pFliC(-gly) were dose-dependent. Lower doses delivered intranasally re-capitulated
the effects seen after systemic and dermal routes giving increases in anti-OVA IgG in the sera as well
as IFNγ-producing Class I-dependent cellular responses. Higher doses of pFliC(-gly) however, were
able to induce mucosal anti-OVA IgG and IgA responses. These observations suggest there may be a
lower threshold for flagellin to promote systemic responses to an antigen compared to mucosal responses,
which might require more of the adjuvant. Whether this could be through the triggering of a threshold
of pre-existing cells at the vaccination site, recruitment of new cell populations to the site after
vaccination, or differences in triggering TLR5 and NLRC4 responses is not known.
3.5. gp160 DNA and Protein Vaccinations; Timeline and Antibody Responses
Experiments with pOVA and pFliC(-gly) indicated that delivery of plasmids using N3 and the
intranasal route was able to promote cellular immune responses as well as humoral mucosal immune
responses. To compare the effectiveness of secreted flagellin to promote immune responses to a clinical
antigen using a heterologus prime/boost regime, priming intranasal DNA vaccinations were carried out
using plasmid pgp160Lfai/pRev [16] with delivery lipid N3 alone or together with pFliC(-gly). Boostings
were performed using recombinant gp160 proteins with a protein-delivery lipid L3B alone or together
with N3 mixed with pFliC(-gly) (Table 2). In these experiments mice were given doses of antigen and
adjuvant believed to maximize detectable responses. Mice were primed, boosted, and analyzed according
to the indicated timeline (Figure 5a). Four weeks post-final boost serum total-IgG titers anti-rgp160
indicated that addition of N3 to pgp160 was able to strongly promote anti-gp160 antibody responses
(Figure 5b). Similar to responses seen using OVA, addition of N3/pFliC(-gly) to the immunization
regime enhanced antigen-specific antibody titers further (Figure 5b). The adjuvant effect was
dependent on N3. Likely due to it’s ability to encapsulate plasmid DNA and protect it from the degradative
environment of the mucosal compartment. These higher titers of antigen-specific IgG in the sera and
the presence of antigen-specific mucosal IgA indicate that the mucosal adjuvant effects of pFliC(-gly)
are not limited to experimental antigens.
Kinetic analysis of anti-gp160 IgG isotypes revealed that addition of N3 to pgp160 was able to
promote anti-gp160 IgG1 at 4 weeks following the final boost. This titer was generally sustained to
24 weeks but fell sharply by week 36 (Figure 5c). A similar trend was seen when anti-gp160 IgG2a was
studied (Figure 5d). When pFliC(-gly) was added to N3/pgp160 vaccinations IgG1 and IgG2a anti-gp160
titers were enhanced further as well as sustained to later time points (Figure 5c,d). These results
suggest that the adjuvant effects of pFliC(-gly) not only boost antigen-specific antibody but that these
effects may also lead to longer persistence of antibody.
Vaccince 2013, 1
430
One should bear in mind that the antigen doses used were chosen to be relatively weak at inducing
immune responses when given via the nasal route in small volume once or twice without adjuvant.
Therefore, it was not surprising to see a short-lived serum and mucosal antibody response unless
adjuvant was used. Longevity of total anti-gp160 IgG was significantly enhanced in groups where the N3
adjuvant was used especially when combined with pFliC-DNA. Since the N3 adjuvant has cationic and
surfactant properties one proposed mechanism would be that there is an increased mucosa-penetrating
property when HIV-antigen and adjuvant is given in mixture. This would increase the amount of
antigen reaching below the mucosal surface, thereby making the antigen available at higher dose for
antigen-presenting cells in the mucosa. Equally important, the capacity of the serum immunoglobulins to
neutralize HIV-1 virus in vitro, both cell-line adapted (HIV-1IIIB), and primary patient isolate (HIV-1
6794B) remained clearly detectable at 36 weeks in serum from groups 2 and 4 (Figure 5e).
Although we observe enhanced titers of anti-viral spike antigen antibodies in the serum of animals
immunized with antigen and pFliC(-gly) adjuvant the antibodies in mucosal secretions may be more
likely to potentially neutralize viral particles. Studies of vaginal IgA anti-gp160 responses had assay
backgrounds that precluded the determination of antigen-specific titers (data not shown). As a surrogate
location representative of antigen-specific IgA responses we studied the titers of IgA anti-gp160
harvested from nasal washes. We observed trends similar to those found in the serum. Addition of N3
to pgp160 vaccinations followed by L3B protein boostings lead to clear IgA anti-gp160 titers which
could be further enhanced by the addition of pFliC(-gly) (Figure 6a). The ability of nasal IgA anti-gp160
to cross-react against homologous (clade B) as well as heterologus clades (A and C) of HIV-1 gp160
was also tested. We observed nasal wash reactivity to clades A and B (Figure 6b). As with serum IgG,
N3 could promote detectable anti-gp160 antibodies while pFliC(-gly) could enhance responses even
further (Figure 6b). These results indicate that higher titers of antigen-specific clade B160 IgA also
correlate with higher titers of antibody able to cross react with HIV-1 clade A gp160. Increases in
cross-clade reactivity may likely be a behavior central to the development of an effective vaccine.
Figure 5. Vaccination schedule, serum antibody responses to gp160, and virus neutralization
titers. (a) Immunization and sample isolation timeline. Priming (ImmunogenP, plasmids) and
boostings (ImmunogenB, rec proteins) are indicated in days while time after the final boost
are indicated in weeks. Immunization details are listed in Table 2; (b) Serum IgG titer against
rgp160 at 4 weeks post immunization in all seven study groups; (c) Serum titer anti-rgp160
IgG1 isotype kinetics in the four first study groups in Table 2; (d) Serum end-point titer
anti-rgp160 IgG2a isotype kinetics in the four first study groups. The concentration of
rgp160-specific Abs are expressed as the end-point titers giving an OD equal to, or higher
than, the mean + 3 SDs (the determined cutoff value for the assay) of the values of serum
samples from unimmunized mice. Absorbance values equal to or above the cutoff value
were considered positive; (e) Serum neutralization of HIV shown as IC50 in serum
samples of the four first study groups in Table 2. The TCID50 (the reciprocal of the virus
dilution where 50% of the cultures were infected) of IIIB (LAI) or 6794 was incubated
with sample mouse serum (dilutions: 20, 60, 180, 540, 1 620). 5 × 104 cells well were then
added, incubated, washed, and incubated for 7 days. Culture supernatants were tested for
virus production by HIV-1 p24 capture ELISA. The lowest serum concentration giving a
Vaccince 2013, 1
50% reduction (IC50) of ELISA absorbance value compared with the mean of the negative
controls are presented [19]. Statistical analyses were conducted using a two-tailed unpaired
Student t test. * Differences of the responses between compared groups defined as p ≤ 0.05
were considered significant. n.s. = non-significant. Comparisons between groups with the
HIV-1 antigens were performed by using the non-parametric Mann-Whitney U test with
Bonferroni correction, p < 0.05 was considered significant.
431
Vaccince 2013, 1
432
Figure 6. Mucosal antibody responses to gp160. (a) Nasal IgA anti-rgp160; (b) Nasal IgA
anti-gp160 cross-reactivity against clade A, B, and C envelope antigens. Priming
(ImmunogenP, plasmids) and boosting (ImmunogenB, rec proteins) groups are shown in
the key. Immunization details are listed in Table 2. ELISA was performed using individual
serum from the indicated immunization groups. Absorbance values equal to or above the
cutoff value were considered positive. Statistical analyses were conducted using a two-tailed
unpaired Student t test. * Differences of the responses between compared groups defined as
p ≤ 0.05 were considered significant.
To determine the breadth of antibody responses against hypervariable regions of gp160 within
antigen-specific IgG we performed B cell epitope mapping of group-pooled serum against individual
20-mers of gp160 from AA249–499 containing the V3 variable loop region. We observed clear populations
of IgG anti-gp160 peptide immune responses in the sera of mice immunized with pgp160 with N3
followed by boosting with rgp160 protein with L3B (Figure 7a). However, the addition of pFliC(-gly)
to immunizations expanded the number of detectable populations by five (Figure 7a). Analysis of
amino acid identities and similarities between FliC and clade B LAI gp160 within the region encoded
by the peptides were performed using NCBI BLASTP analysis (v2.2.26+) with default settings. Two
regions were identified containing identity and similarity. Of these two regions only one (containing
Vaccince 2013, 1
433
36% identities and 55% similarity) overlapped with a region of increased reactivity (peptides AA 439
and 444) and was excluded. There were no regions of alignment identified within the 5 annotated
populations that exhibited equal or greater identity and similarity than the 22AA region.
Figure 7. B cell epitope mapping to C2-C5 region of gp160 after immunization with gp160
with and without adjuvant. ELISA was performed using group-pooled serum (equal
volumes) from immunization group 2 (n = 35) or 4 (n = 35) against individual peptides.
Priming (ImmunogenP, plasmids) and boosting (ImmunogenB, rec proteins) groups are shown
in the key. Immunization details are listed in Table 2. The concentration of gp160-peptide
specific Abs are expressed as the end-point titers giving an OD equal to, or higher than, the
mean + 3 SDs (the determined cutoff value for the assay) of the values of serum samples
from unimmunized mice. Absorbance values equal to or above the cutoff value were
considered positive. Statistical analyses were conducted using a two-tailed unpaired
Student t test. * Differences of the responses between compared groups defined as p ≤ 0.05
were considered significant.
These use of pFliC(-gly) appears to promote a broadening of B cell epitope reactivity to rgp160
and/or presentation of additional ―masked‖ epitopes. Similar responses have been seen in response to a
TLR-adjuvanted malaria vaccine using advanced techniques [26]. However, how this increased reactivity
occurs is not known. It may be that the higher titers of anti-gp160 elicited by use of pFliC(-gly) revealed
reactivity normally below threshold when samples from un-adjuvanted groups were studied. Additionally,
it may be that pFliC(-gly) is able to promote expansion of B cell populations normally under
stimulated or neglected due to competition. However, it may also be possible that there is cross-epitope
reactivity between rgp160 and FliC, and new regions of anti-gp160 reactivity are actually due to anti-FliC
antibodies. Although this cannot be formally excluded here it may be unlikely due to the low nature of
homology between antigen (gp160) and adjuvant (FliC). Nevertheless, these results suggest that
detailed study of antibody responses in mice receiving pFliC(-gly) are warranted.
3.6. gp160 and p24gag DNA and Protein Vaccinations; Cellular Responses
To study T cell immune responses to DNA-prime/protein-boost i.na. immunizations we chose to
assay standard cytokines associated with Th1-like (IFN) or Th2-like (IL-5) populations. Responses to
Vaccince 2013, 1
434
gp160 were assayed following individual splenocytes harvesting at 4, 8, 12, 24, and 36 weeks after
final boost and restimulation with rgp160. Observed response trends were similar to those seen when
studying antibody responses. Addition of N3 to pgp160 vaccinations followed by L3B protein
boostings lead to clear and significant IFN, IL-5, and proliferative responses over mice immunized
without adjuvant (Figure 8a–c). The IFN and proliferative responses could be further enhanced by the
addition of pFliC(-gly) but not IL-5 (Figure 8a–c) demonstrating the ability of pFliC(-gly) to act as an
adjuvant but with a propensity to strengthen Th1-like responses.
To study the T cell immune responses to p24gag individual splenocytes were harvested at 4 weeks after
final boost and restimulated with rp24gag. Interestingly, the addition of N3 to p24gag vaccinations
followed by L3B protein boostings only lead to clear and significant increases in IL-5 and proliferative
responses compared to mice immunized without adjuvant (Figure 9a–c). Increases in IFN production were
not seen. IFN and proliferative responses could be significantly enhanced by the addition of pFliC(-gly)
but the IL-5 responses gained by use of N3 were suppressed by the addition of pFliC(-gly) (Figure 9a–c).
Interestingly the abilities of N3 and pFliC(-gly) to act as adjuvants did not completely overlap and,
in one combination, even counteracted the other. In our immunizations the secreted antigen gp160 with
N3 promoted a somewhat Th2-like response (including IFN) which was further enhanced by the
addition of pFliC(-gly). Similar results were seen with the intracellular antigen p24gag where addition
of pFliC(-gly) promoted IFN responses [27–31]. However, with p24gag the pFliC(-gly) addition
actually suppressed IL-5 responses.
Together these results suggest that N3 has the ability to promote Th2-like adjuvant effects (IL-5 and
proliferation) to extracellular and intracellular antigens whereas the effects of pFliC(-gly), which were
greatly dependent on the presence N3, promoted Th1-like adjuvant effects (IFN, proliferation)
sometimes at the expense of Th2-like responses (IL-5). Why these effects were dependent on the
―location‖ of the antigen is unknown. However, it does suggest that with complex antigen/adjuvant
mixtures that we are unable to predict exactly how they will affect adaptive immune responses.
3.7. Immune Activation Potential of Adjuvants
To determine the types of inflammatory factors elicited intranasally by the adjuvants used we
performed nasal mucosal washings at various time points after adjuvant delivery. We detected
increases in the inflammatory cytokines IL-6, IFN, and IFN at 18 to 48 h post-nasal adjuvant
administration compared to nasal saline exposure (Figure 10a–c). Significant increases in IL-6 and
IFN were elicited by N3 use at 18 hours post exposure while significant increases in IL-6 was elicited
using a combination of pFliC(-gly) and N3 only at later time points (Figure 10a). These increases in
IL-6 were dependent on the use of N3 with pFliC(-gly) but were not due to N3 or pFliC(-gly) alone.
3.8. Longevity of Immune Responses
Currently, it is still likely that several alternative prime-boost combinations will need to be tested to
identify the most promising vaccine/adjuvant and vaccine design regimes for HIV-1 vaccines [32–34].
Vaccince 2013, 1
Figure 8. Kinetic analysis of T cell responses to immunizations with gp160 with and
without adjuvant combinations. (a) Anti-mIFN ELISA was performed on cells restimulated
with rgp160. Values shown were adjusted for baseline values seen using identical stimulations
using splenocytes from naive mice. Priming (ImmunogenP, plasmids) and boosting
(ImmunogenB, rec proteins) groups are shown in the key. Immunization details are listed
in Table 2; (b) Anti-mIL-5 ELISA was performed on cells restimulated with rgp160.
Values shown were adjusted for baseline values seen using identical stimulations using
splenocytes from naïve mice; (c) Proliferative response to stimulation with rgp160 defined
as stimulation index relative to identical stimulations using splenocytes from naïve mice.
Statistical analyses were conducted using a two-tailed unpaired Student t test. * Differences
of the responses between compared groups at week 4 after final boost defined as p ≤ 0.05
were considered significant. n.s. = non-significant. Comparisons between groups with the
HIV-1 antigens were performed by using the non-parametric Mann-Whitney U test with
Bonferroni correction, p < 0.05 was considered significant.
435
Vaccince 2013, 1
Figure 9. Analysis of T cell responses to immunizations with p24gag with and without
adjuvant combinations. (a) Anti-mIFN ELISA was performed on cells restimulated with
p24gag. Values shown were adjusted for baseline values seen using identical stimulations
using splenocytes from naïve mice. Priming (ImmunogenP, plasmids) and boosting
(ImmunogenB, rec proteins) groups are shown in the key. Immunization details are listed
in Table 2; (b) Anti-mIL-5 ELISA was performed on cells restimulated with p24gag.
Values shown were adjusted for baseline values seen using identical stimulations using
splenocytes from naïve mice; (c) Proliferative response to stimulation with p24gag defined
as stimulation index relative to identical stimulations using splenocytes from naïve mice.
Statistical analyses were conducted using a two-tailed unpaired Student t test. * Differences
of the responses between compared groups at week 4 after final boost defined as p ≤ 0.05
were considered significant. n.s. = non-significant. Comparisons between groups with the
HIV-1 antigens were performed by using the non-parametric Mann-Whitney U test with
Bonferroni correction, p < 0.05 was considered significant.
436
Vaccince 2013, 1
437
Figure 10. Cytokines produced after intranasal adminstration of adjuvant combinations.
Kinetic analysis of (a) IL-6, (b) IFN, and IFN2 (c) at 18, 36, and 48 h by ELISA using
nasal wash samples. Mock adjuvant shown as White bars, use of pFliC(-gly) as Striped
Bars, and N3 as Grey bars (n = 5 mice/group). Data is expressed as the calculated
mean ± SEM. Statistical analyses were conducted using a two-tailed Mann-Whitney test.
** Differences relative compared groups defined as p ≤ 0.005 were considered significant.
There are several important aspects concerning the flexibility associated with HIV-vaccine antigen
development: first, the selection of immunogens and adjuvants. In this study, and in several others,
DNA-plasmids should be selected that have long-lasting stability and allow persistence as stable
antigen-expressing vectors. Second, production of DNA-plasmids, is today quite efficacious and can easily
be performed at large-scale. Third, DNA-plasmids are attractive due to their great adaptability, and
modifications in expression efficacy, gene exchange, or other desired modification is today easy to carry out.
Recombinant HIV-proteins as vaccine antigens are instead more of a challenge, especially when it
comes to production and expression of such delicate proteins like the HIV-1 outer envelope. If they need
to structurally mimic the envelope spikes found at the surface of HIV-1 primary isolates they need to be
produced and maintained as multimeric, glycosylated envelope proteins. This production is not a trivial
matter, and search for the ideal HIV-1 envelope vaccine candidate is still an unsolved issue [32,33].
Finally, the stable storage of sensitive recombinant proteins antigens is likely more of a problem. Thus
a potent and safe adjuvant, formulated with obtainable amounts of quality antigen, may be a critical
way to use these immunogenic proteins as vaccine candidates. In this study, we chose the recombinant
baculovirus expressed HIV-1 gp160 due to its modest immunogenicity, its fair degree of glycosylation
and trimeric structure and the content of both gp41 and gp120 envelope proteins [16,18].
From an immunological perspective, immunizing with a DNA plasmid with its endogenous in vivo
expression of the HIV-antigen and especially of a highly glycosylated, conformation-sensitive antigens
as HIV-1 gp120 is an attractive technology. Much of the trouble of production, safe storage and
efficient administration of a neutralization antibody-inducing multimeric protein may then be reduced
or avoided [35].
The durability of protective immune responses can often be enhanced by broadening antigen
recognition ability through enhanced antigen delivery, enhanced antigen uptake, and/or prolonged
antigen exposure [36,37]. Furthermore, by triggering several innate immune recognition pathways such
as multiple Toll-like receptors (with DNA-plasmid CpG motifs, (TLR9), with FliC expression (TLR5),
cytoplasmic DNA sensors [38,39] and increased induction of cell death using surfactant adjuvants in
Vaccince 2013, 1
438
conjunction with antigen presentation as with cationic lipids more potent immune enhancement may be
obtained [40]. In this study, we endeavored to optimize the immunological outcome (strength and
longevity) of these vaccine technologies by methodologically studying optimal route delivery for
adjuvants with DNA-plasmid vaccination followed by the application of this knowledge to a heterologus
prime-boost regime using HIV-1 antigens. Importantly, we show that in the HIV-1 study group (group
No.4) receiving potentially the broadest HIV-1 antigen variants and the most complex adjuvant
combination we obtained the most long-lasting humoral and cell mediated immune responses.
3.9. Impact of Adjuvant on Local Innate Immunity
Certain studies have described the induction of innate and adaptive immune reactivity when
immunogens and adjuvant have been given to mice [41–43]. In the current studies we have performed
analyses on how the cationic lipid-based N3 adjuvant, the DNA-plasmid-based FliC (-gly) adjuvant,
and the combination of both together influence the local and systemic innate and adaptive immune
responses. The data indicate that the adjuvants stimulate the production of pro-inflammatory IL-6 and
interferon (IFN), two cytokines described promote innate immunity and B cell activation [44,45].
Each of the adjuvants are likely to contribute, in their own way, to immune activation, and lipid-based
adjuvants (such as the N3) have been observed to induce cell death (as seen with N3 in vitro, data not
shown) and local inflammation. This would provide danger signals to attract antigen-presenting cells,
stimulate antigen uptake, and induce dendritic cell maturation. For instance, cell death and endogenous
DNA release would be able to function as an endogenous adjuvant capable of supporting IL-6 release
and a T-helper type 2 response [41,45,46]. Inflammatory cytokine patterns, accompanied higher
antibody titers have been reported by Valesi et al., when using the MF59 lipid emulsion adjuvant and
influenza vaccine given to mice [47]. Similar results in inflammatory cytokine production with the N3
lipid adjuvant and HIV antigens were also observed in our work.
DNA-plasmid vaccination used to express antigen and adjuvant proteins can trigger TLR systems and
promote inflammatory cytokine production (such as IL-6) [48]. Intranasal recombinant FliC polypeptide
has also been observed to induce inflammatory cytokines including IL-6 [49]. Interestingly, the
induction of IL-6 secretion in mucosal stimulation has been reported to improve transepithelial
transport over mucosal epithelial barriers [50]. This effect of IL-6 may explain the enhanced mucosal
immune responses we observe using mucosally delivered pFliC(-gly). However, the ability of FliC to
promote mucosal adaptive immunity is complex and appears to also involve the production of
numerous other factors not studied here such as IL-17, IL-22, and IL-23 [51,52].
Our observations of mucosal, antigen-specific IgA elicited by intra-nasal immunization indicates
presence of a Th2-type response with N3 use while IFN production detected in the nasal washes of
pFliC(-gly) immunized animals suggest a Th1-type response. Our results also suggest that these
responses need not be mutually exclusive. Adjuvant emulsions such as MF59 and similar products
applied mucosally have been shown to act as Th2-type adjuvants, and addition of additional stimuli such
as CpGs, and other TLR agonists have been observed to skew immune responses towards a Th1-type or
a balanced Th1/Th2-type immune pattern [38,53]. Here, we show data that immune responses induced
using a Th2-enhancing lipid adjuvant (N3) may be modified by the addition of a TLR-agonist and
inflammasome trigger (pDNA, FliC), to also promote a Th1-type (IFN) response when administered nasally.
Vaccince 2013, 1
439
4. Conclusions
Adjuvant choice during DNA vaccine development will depend on formulation relative to method
of delivery, the recipient, the protective antigens used, as well as the desired induction of immunity at
the portal of infection. As the detection of flagellin by innate immune receptors is evolutionarily
conserved, it has the potential to be easily used in many species without the need to isolate and prepare
species-specific adjuvants such as cytokines [27–31]. These unique properties as well as its ability to
promote both humoral and cellular responses to co-delivered antigens by multiple routes without a
need for manipulating the antigen indicate that it works as an easy and efficient adjuvant to improve
non-living non-replicating DNA vaccines.
Finally, in this work, all immunizations with HIV-antigens were delivered as heterologous prime
boost immunizations. Antigens are presented both through endogenous expression and as recombinant
soluble proteins to the immune system to provide the antigenic regions or epitopes with greater
variation than in a homologous immunization. With this approach we demonstrate that cationic lipids
formulated with plasmid FliC-DNA and plasmid HIV-DNA, followed by cationic lipids formulated
with plasmid FliC-DNA and recombinant HIV-1 proteins (study group 4) elicited the longest-lived
immunity and broadest antigen epitope recognition.
Acknowledgments
This work was supported by the Swedish International Development Cooperation Agency (SIDA),
the Swedish Research Council (VR), and Läkare mot AIDS research fund. We would like to thank
Lech Ignatowitz for fresh N3 preparation, Eurocine Vaccines AB for providing N3 and L3B adjuvants.
David Hallengärd for excellent ELISPOT assistance.
Conflicts of Interest
Jorma Hinkula and Ulf Schröder have IP-rights and economical interests in the N3 adjuvant. All
other authors have no conflicts to declare.
References
1.
2.
3.
4.
5.
Jones, S.; Evans, K.; McElwaine-Johnn, H.; Sharpe, M.; Oxford, J.; Lambkin-Williams, R.; Mant, T.;
Nolan, A.; Zambon, M.; Ellis, J.; et al. DNA vaccination protects against an influenza challenge
in a double-blind randomised placebo-controlled phase 1b clinical trial. Vaccine 2009, 27, 2506–2512.
Girard, M.P.; Bansal, G.P. HIV/AIDS vaccines: A need for new concepts? Int. Rev. Immunol.
2008, 27, 447–471.
Jechlinger, W. Optimization and delivery of plasmid DNA for vaccination. Expert Rev. Vaccines
2006, 5, 803–825.
Miao, E.A.; Andersen-Nissen, E.; Warren, S.E.; Aderem, A. TLR5 and Ipaf: Dual sensors of
bacterial flagellin in the innate immune system. Semin. Immunopathol. 2007, 29, 275–288.
Applequist, S.E.; Rollman, E.; Wareing, M.D.; Liden, M.; Rozell, B.; Hinkula, J.; Ljunggren, H.G.
Activation of innate immunity, inflammation, and potentiation of DNA vaccination through
mammalian expression of the TLR5 agonist flagellin. J. Immunol. 2005, 175, 3882–3891.
Vaccince 2013, 1
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
440
Song, L.; Nakaar, V.; Kavita, U.; Price, A.; Huleatt, J.; Tang, J.; Jacobs, A.; Liu, G.; Huang, Y.;
Desai, P.; et al. Efficacious recombinant influenza vaccines produced by high yield bacterial
expression: A solution to global pandemic and seasonal needs. PLoS One 2008, 3, e2257.
Mizel, S.B.; Graff, A.H.; Sriranganathan, N.; Ervin, S.; Lees, C.J.; Lively, M.O.; Hantgan, R.R.;
Thomas, M.J.; Wood, J.; Bell, B. Flagellin-F1-V fusion protein is an effective plague vaccine in
mice and two species of nonhuman primates. Clin. Vaccine Immunol. 2009, 16, 21–28.
Mizel, S.B.; Bates, J.T. Flagellin as an adjuvant: Cellular mechanisms and potential. J. Immunol.
2010, 185, 5677–5682.
Lycke, N. Recent progress in mucosal vaccine development: Potential and limitations. Nat. Rev.
Immunol. 2012, 12, 592–605.
Chen, W.; Patel, G.B.; Yan, H.; Zhang, J. Recent advances in the development of novel mucosal
adjuvants and antigen delivery systems. Hum. Vaccine 2010, 6, 706–714.
Hinkula, J.; Bratt, G.; Gilljam, G.; Nordlund, S.; Broliden, P.A.; Holmberg, V.; Olausson-Hansson, E.;
Albert, J.; Sandstrom, E.; Wahren, B. Immunological and virological interactions in patients
receiving passive immunotherapy with HIV-1 neutralizing monoclonal antibodies. J. Acq.
Immune Defic. Syndr. 1994, 7, 940–951.
Sutlu, T.; Nystrom, S.; Gilljam, M.; Stellan, B.; Applequist, S.E.; Alici, E. Inhibition of intracellular
anti-viral defense mechanisms augments lentiviral transduction of human natural killer cells:
Implications for gene therapy. Hum. Gene Ther. 2012, 23, 1090–1100.
Hinkula, J.; Devito, C.; Zuber, B.; Benthin, R.; Ferreira, D.; Wahren, B.; Schroder, U. A novel
DNA adjuvant, N3, enhances mucosal and systemic immune responses induced by HIV-1 DNA
and peptide immunizations. Vaccine 2006, 24, 4494–4497.
Petersson, P.; Hedenskog, M.; Alves, D.; Brytting, M.; Schroder, U.; Linde, A.; Lundkvist, A.
The Eurocine L3 adjuvants with subunit influenza antigens induce protective immunity in mice
after intranasal vaccination. Vaccine 2010, 28, 6491–6497.
Meyers, G.; Korber, B.; Foley, K.T.; Jeang, J.W.; Mellers, J.W.; Wain-Hobson, S. A Compilation
and Analysis of Nucleic Acid and Amino Acid Sequences; Los Alamos, Los Alamos National
Laboratory: Washington, DC, USA, 1992.
Brave, A.; Hallengard, D.; Schroder, U.; Blomberg, P.; Wahren, B.; Hinkula, J. Intranasal
immunization of young mice with a multigene HIV-1 vaccine in combination with the N3
adjuvant induces mucosal and systemic immune responses. Vaccine 2008, 26, 5075–5078.
Hinkula, J.; Hagbom, M.; Wahren, B.; Schroder, U. Safety and immunogenicity, after nasal
application of HIV-1 DNA gagp37 plasmid vaccine in young mice. Vaccine 2008, 26, 5101–5106.
Hinkula, J.; Rollman, E.; Lundholm, P.; Benthin, R.; Okuda, K.; Wahren, B. Genetic immunization
with multiple HIV-1 genes provides protection against HIV-1/MuLV pseudovirus challenge in vivo.
Cells Tissues Organs 2004, 177, 169–184.
Devito, C.; Levi, M.; Broliden, K.; Hinkula, J. Mapping of B-cell epitopes in rabbits immunised
with various gag antigens for the production of HIV-1 gag capture ELISA reagents. J. Immunol.
Methods 2000, 238, 69–80.
Lightfield, K.L.; Persson, J.; Brubaker, S.W.; Witte, C.E.; von Moltke, J.; Dunipace, E.A.; Henry, T.;
Sun, Y.H.; Cado, D.; Dietrich, W.F.; et al. Critical function for Naip5 in inflammasome activation
by a conserved carboxy-terminal domain of flagellin. Nat. Immunol. 2008, 9, 1171–1178.
Vaccince 2013, 1
441
21. Nystrom, S.; Antoine, D.J.; Lundback, P.; Lock, J.G.; Nita, A.F.; Hogstrand, K.; Grandien, A.;
Erlandsson-Harris, H.; Andersson, U.; Applequist, S.E. TLR activation regulates damage-associated
molecular pattern isoforms released during pyroptosis. EMBO J. 2013, 32, 86–99.
22. Takeshita, F.; Ishii, K.J. Intracellular DNA sensors in immunity. Curr. Opin. Immunol. 2008, 20,
383–388.
23. Schroder, K.; Muruve, D.A.; Tschopp, J. Innate immunity: Cytoplasmic DNA sensing by the
AIM2 inflammasome. Curr. Biol. 2009, 19, R262–R265.
24. Uematsu, S.; Fujimoto, K.; Jang, M.H.; Yang, B.G.; Jung, Y.J.; Nishiyama, M.; Sato, S.; Tsujimura, T.;
Yamamoto, M.; Yokota, Y.; et al. Regulation of humoral and cellular gut immunity by lamina
propria dendritic cells expressing Toll-like receptor 5. Nat. Immunol. 2008, 9, 769–776.
25. Atif, S.M.; Uematsu, S.; Akira, S.; McSorley, S.J. CD103-CD11b+ dendritic cells regulate
the sensitivity of CD4 T-cell responses to bacterial flagellin. Mucosal. Immunol. 2013,
doi:10.1038/mi.2013.25.
26. Wiley, S.R.; Raman, V.S.; Desbien, A.; Bailor, H.R.; Bhardwaj, R.; Shakri, A.R.; Reed, S.G.;
Chitnis, C.E.; Carter, D. Targeting TLRs expands the antibody repertoire in response to a malaria
vaccine. Sci. Transl. Med. 2011, doi:10.1126/scitranslmed.3002135.
27. Vajdy, M.; Srivastava, I.; Polo, J.; Donnelly, J.; O’Hagan, D.; Singh, M. Mucosal adjuvants and
delivery systems for protein-, DNA- and RNA-based vaccines. Immunol. Cell. Biol. 2004, 82,
617–627.
28. Toka, F.N.; Pack, C.D.; Rouse, B.T. Molecular adjuvants for mucosal immunity. Immunol. Rev.
2004, 199, 100–112.
29. Han, T.K.; Dao, M.L. Enhancement of salivary IgA response to a DNA vaccine against Streptococcus
mutans wall-associated protein A in mice by plasmid-based adjuvants. J. Med. Microbiol. 2007,
56, 675–680.
30. Kataoka, K.; McGhee, J.R.; Kobayashi, R.; Fujihashi, K.; Shizukuishi, S. Nasal Flt3 ligand cDNA
elicits CD11c+CD8+ dendritic cells for enhanced mucosal immunity. J. Immunol. 2004, 172,
3612–3619.
31. Melkebeek, V.; Sonck, E.; Verdonck, F.; Goddeeris, B.M.; Cox, E. Optimized FaeG expression
and a thermolabile enterotoxin DNA adjuvant enhance priming of an intestinal immune response
by an FaeG DNA vaccine in pigs. Clin. Vaccine Immunol. 2007, 14, 28–35.
32. Van Gils, M.J.; Sanders, R.W. Broadly neutralizing antibodies against HIV-1: Templates for a
vaccine. Virology 2013, 435, 46–56.
33. Girard, M.P.; Osmanov, S.K.; Kieny, M.P. A review of vaccine research and development: The
human immunodeficiency virus (HIV). Vaccine 2006, 24, 4062–4081.
34. McGuire, A.T.; Hoot, S.; Dreyer, A.M.; Lippy, A.; Stuart, A.; Cohen, K.W.; Jardina, J.; Menis, S.;
Scheid, J.F.; West, A.P.; et al. Engineering HIV envelope protein to activate germline B cell receptors
of broadly neutralizing anti-CD4 binding site antibodies. J. Exp. Med. 2013, 210, 656–663.
35. Narayan, K.M.; Agrawal, N.; Du, S.X.; Murakana, J.E.; Bauer, K.; Leaman, D.P.; Phung, P.;
Limoli, K.; Chen, H.; Boenig, R.I.; et al. Prime-boost immunization of rabbits with HIV-1 gp120
elicits potent neutralizing activity against a primary viral isolate. PLoS One 2013, 8, e52732.
Vaccince 2013, 1
442
36. Naito, S.; Maeyama, J.-I.; Mizukami, T.; Takahashi, M.; Hamaguchi, I.; Yamaguchi, K. Transcutaneous
immunization by merely prolonging the duration of antigen presence in the skin of mice induces a
potent antigen-specific immune response even in the absence of an adjuvant. Vaccine 2007, 25,
8762–8770.
37. Scharton-Kersten, T.; Glenn, G.M.; Vassell, R.; Yu, J.; Walwender, D.; Alving, C.R. Principles of
transcutaneous immunization using cholera toxin as an adjuvant. Vaccine 1999, 17, S37–S43.
38. Kasturi, S.P.; Skountzou, I.; Albrecht, R.A.; Koutsonanos, D.; Hua, T.; Nakaya, H.I.; Ravindran, R.;
Stewart, S.; Alam, M.; Kwissa, M.; et al. Programming the magnitude and persistence of antibody
responses with innate immunity. Nature 2011, 470, 543–547.
39. Napoliatani, G.; Rinaldi, A.; Bertoni, F.; Sallusto, F.; Lanzavecchia, A. Selected Toll-like receptor
agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells.
Nat. Immunol. 2005, 6, 769–776.
40. Shen, S.-S.; Yang, Y.-W. Antigen delivery for cross-priming via emulsion vaccine adjuvants.
Vaccine 2012, 30, 1560–1571.
41. Marichal, T.; Ohata, K.; Bedoret, D.; Mesnil, C.; Sabatel, C.; Kobiyama, K.; Lekeux, B.; Coban, C.;
Ishii, K.J.; Bureau, F.; et al. DNA released from dying host cells mediate aluminium adjuvant
activity. Nat. Med. 2011, 17, 996–1003.
42. Yang, Y.-W.; Wei, A.-C.; Shen, S.-S. The immunogenicity-enhancing effect of emulsion vaccine
adjuvant independent of the dispersion type and antigen release rate—A revisit of the role of the
hydrophile-lipophile balance (HLB) valus. Vaccine 2005, 23, 2665–2675.
43. O’Hagan, D.T.; Ott, G.S.; van Nest, G.; Rappuoli, R.; del Guidice, G. The history of M59 adjuvant:
A phoenix that rose from the ashes. Exp. Rev. Vaccines 2013, 12, 13–30.
44. Awasti, A.; Kuchroo, V.K. Th17 cells: From precursors to players in inflammation and infection.
Int. Immunol. 2009, 20, 489–498.
45. Makidon, P.E.; Belyakov, I.M.; Blanco, L.P.; Janczak, K.W.; Landers, J.; Bielinska, A.U.;
Groom, J.V., 2nd; Naker, J.R., Jr. Nanoemulsion mucosal adjuvant uniquely activates cytokine
production by nasal epithelium and induces dendritic cell trafficing. Eur. J. Immunol. 2012, 42,
2073–2086.
46. Albert, M.L.; Sauter, B.; Bhardwaj, N. Dendritic cells aquire antigen from apoptotic cells and
induce class I-restricted CTLs. Nature 1998, 392, 86–89.
47. Valensi, J.P.; Carlson, J.R.; van Nest, G.A. Systemic cytokine profiles in BALB/c mice immunized
with trivalent influenza vaccine containing MF59 oil emulsion and other advanced adjuvants.
J. Immunol. 1994, 153, 4029–4039.
48. Van Duin, D.; Medzhitov, R.; Shaw, A.C. Triggering TLR signaling in vaccination. Trends Immunol.
2006, 27, 49–55.
49. Honko, A.N.; Mizel, S.B. Mucosal administration of flagellin induces innate immunity in the
mouse lung. Infect. Immun. 2004, 72, 6676–6679.
50. Pritts, T.; Hungness, E.; Wang, Q.; Robb, B.; Hershko, D.; Hasselgren, P.O. Mucosal and enterocyte
IL-6 production during sepsis and endotoxemia-role of transcription factors and regulation of
stress response. Am. J. Surg. 2002, 183, 372–383.
Vaccince 2013, 1
443
51. Kinnebrew, M.A.; Buffie, C.G.; Diehl, G.E.; Zenewicz, L.A.; Leiner, I.; Hohl, T.M.; Flavelli, R.A.;
Littman, D.R.; Pamer, E.G. Interleukin 23 production by intestinal CD103(+)CD11b(+) dendritic
cells in response to bacterial flagellin enhances mucosal innate immune defence. Immunity 2012,
36, 276–287.
52. Van Maele, L.; Carnoy, C.; Cayet, D.; Songhet, P.; Dumoutier, L.; Ferrero, I.; Janot, L.; Erard, F.;
Bertout, J.; Leger, H.; et al. TLR5 signaling stimulates the innate production of IL-17 and IL-22
by CD3(neg)CD127+ immune cells in spleen and mucosa. J. Immunol. 2010, 185, 1177–1185.
53. Krieg, A.M. CpG still rocks update on an accidental drug. Nucleic Acid Ther. 2012, 22, 77–89.
© 2013 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article
distributed under the terms and conditions of the Creative Commons Attribution license
(http://creativecommons.org/licenses/by/3.0/).